Elsevier

Brain Research

Volume 1514, 13 June 2013, Pages 18-39
Brain Research

Research Report
An update on the cognitive impact of clinically-used hormone therapies in the female rat: Models, mazes, and mechanisms

https://doi.org/10.1016/j.brainres.2013.01.016Get rights and content

Abstract

In women, ovarian hormone loss associated with menopause has been related to cognitive decline. Hormone therapy (HT) may ameliorate some of these changes. Understanding the cognitive impact of female steroids, including estrogens, progestogens, and androgens, is key to discovering treatments that promote brain health in women. The preclinical literature has presented elegant and methodical experiments allowing a better understanding of parameters driving the cognitive consequences of ovarian hormone loss and HT. Animal models have been a valuable tool in this regard, and will be vital to future discoveries. Here, we provide an update on the literature evaluating the impact of female steroid hormones on cognition, and the putative mechanisms mediating these effects. We focus on preclinical work that was done with an eye toward clinical realities. Parameters that govern the cognitive efficacy of HT, from what we know thus far, include but are not limited to: type, dose, duration, and route of HT, age at HT initiation, timing of HT relative to ovarian hormone loss, memory type examined, menopause history, and hormone receptor status. Researchers have identified intricate relationships between some of these factors by studying their individual effects on cognition. As of late, there is increased focus on studying interactions between these variables as well as multiple hormone types when administered concomitantly. This is key to translating preclinical data to the clinic, wherein women typically have concurrent exposure to endogenous ovarian hormones as well as exogenous combination HTs, which include both estrogens and progestins. Gains in understanding the parameters of HT effects on cognition provide exciting novel avenues that can inform clinical treatments, eventually expanding the window of opportunity to optimally enhance cognition and brain health in aging women.

This article is part of a Special Issue entitled Hormone Therapy.

Section snippets

Ovarian hormones and cognition in the rodent: Historical context and clinical Implications

In 1927, A.S. Parkes published a monograph in The Proceedings of the Royal Society of Medicine entitled “Internal Secretions of the Ovary”, in which it is stated that, “The solution of the type of problem found in studying the internal secretions of the ovary is most satisfactorily sought by experiment, and since the lower mammals have to be used for this type of work, it is on their reactions that our knowledge of ovarian activity mainly depends. At the same time, however, ovarian activity in

Operationally defining and testing memory effects of female steroids in the preclinical setting

When studying learning and memory in the rodent model, it is vital to acknowledge the multiple parameters involved in the process of quantifying cognition in order to properly interpret data. Within the specific domain of spatial navigation, rodents learn to navigate through a novel environment so that routes to the target eventually become familiar, and associations are formed from cues in the environment to aid overall navigation. Spatial learning and memory involves the ability to navigate

Activational versus organizational effects of hormones: Perspectives for discussion herein

Gonadal steroid hormone actions are traditionally referred to as having organizational effects, operationally defined in the traditional sense as occurring early in development and having permanence, or as having activational effects which occur later in development, are transitory, and therefore depend on presence of the hormone at the time of assessment. For example, sex differences in neuroanatomy have been traditionally thought to reflect the permanent organizing effects of steroids present

Menopause etiology

Studies have shown that ovarian hormone loss negatively impacts cognition in women, and that these effects correspond to the associated estrogen deficiency (Farrag et al., 2002, Phillips and Sherwin, 1992, Sherwin, 1988). Others have shown modest, but statistically significant, declines in cognitive performance in women after surgical menopause, but express that the effects were small and not likely to be of clinical significance (Kritz-Silverstein and Barrett-Connor, 2002). In numerous

Estrogens

Estrogens are a class of hormones including 17β-estradiol, estrone, and estriol. 17β-estradiol is the most potent naturally-circulating estrogen, followed by estrone and estriol, in order of receptor affinity (Kuhl, 2005, Sitruk-Ware, 2002). Since the first controlled clinical evaluation showing that 17β-estradiol injections enhanced memory in 75 year-old women (Caldwell and Watson, 1952), numerous studies have demonstrated cognitive decline after ovarian hormone loss, and enhancement after

Progestogens

Progestogens include steroids with a pregnane skeleton, including naturally-occurring progesterone as well as progestins (synthetic progesterones). Inherent to any discussion on the impact of HT on cognition and the complexities involved in outcome, is that of combination therapy, which includes estrogen plus a progestogen concomitantly. Investigating combination therapy is crucial since women with a uterus taking estrogens must include a progestogen in their regimen to offset the increased

Androgens

Androgens are typically thought of as a “male” hormone; a masculinizing hormone which initiates permanent organizational effects on the male brain during a specific critical period in early development, and an activating hormone for male sex behaviors in adulthood. Why study androgens in females as a potential modulator of learning and memory? From our perspective, studying the impact of androgens on cognition in the female rodent model is important for several reasons. First, androgens bind to

Gonadotropins

Although it is well established that the gonadotropins (peptide hormones released from the anterior pituitary) follicle stimulating hormone (FSH) and lutenizing hormone (LH) are involved in regulating reproductive functions via negative and positive feedback loops, increasing evidence is indicating that gonadotropins, directly or indirectly, impact cognitive function as well, including within the spatial domain. Although the links between FSH and cognition do not appear to be realized (e.g.,

Cholinergic and γ-aminobutyric acid (GABA)ergic systems

Abundant evidence suggests that the cholinergic and GABAergic systems are intimately related to the effects of female steroids on cognition. Pharmacological experiments using both peripheral and intracranial infusions show that the cholinergic system may mediate estrogen-induced effects. Much of the landmark work evaluating relations between estrogens and the cholinergic system has been done via the creative and methodical approach of combining hormones and pharmaceutical agents in the

Hormone receptors and the cognitive effects of hormones

The cognitive effects of steroid hormones discussed thus far are, in part, mediated by distinct expression of receptors in the brain. Since its discovery in 1966, estrogen receptor-alpha (ERα) was the first, and thought to be the only, member of the nuclear receptor superfamily that exhibites specificity for 17β-estradiol (Toft and Gorski, 1966). It was not until 30 years later that the second ER subtype, ERβ, was discovered (Kuiper et al., 1996). The two ERs share a significant degree of

General conclusions

Gonadal hormones have activational effects on cognition. Interest in elucidating the factors that mediate these effects has intensified over the last decade. Notably, this heightened interest is within the realm of both the depth and breadth of the impact that ovarian hormones might exert on cognition. Of these effects, the significance of age-related cognitive decline, and how it may be exacerbated by concurrent gonadal hormone loss, has been an increasingly prominent area of focus in female

References (240)

  • V. Birzniece et al.

    GABA(A) receptor changes in acute allopregnanolone tolerance

    Eur. J. Pharmacol.

    (2006)
  • R.L. Bowen et al.

    Luteinizing hormone, a reproductive regulator that modulates the processing of amyloid-beta precursor protein and amyloid-beta deposition

    J. Biol. Chem.

    (2004)
  • B.B. Braden et al.

    Medroxyprogesterone acetate impairs memory and alters the GABAergic system in aged surgically menopausal rats

    Neurobiol. Learn Mem.

    (2010)
  • M. Brett et al.

    Motherhood and memory: a review

    Psychoneuroendocrinology

    (2001)
  • S. Campbell et al.

    Oestrogen therapy and the menopausal syndrome

    Clin. Obstet. Gynaecol

    (1977)
  • G. Casadesus et al.

    Luteinizing hormone modulates cognition and amyloid-beta deposition in Alzheimer APP transgenic mice

    Biochim. Biophys. Acta.

    (2006)
  • G. Casadesus et al.

    Increases in luteinizing hormone are associated with declines in cognitive performance

    Mol. Cell Endocrinol.

    (2007)
  • E.J. Chesler et al.

    Acute administration of estrogen and progesterone impairs the acquisition of the spatial morris water maze in ovariectomized rats

    Horm. Behav

    (2000)
  • L.H. Coker et al.

    Postmenopausal hormone therapy and cognitive outcomes: the Women's Health Initiative Memory Study (WHIMS)

    J. Steroid. Biochem. Mol. Biol.

    (2010)
  • C.D. Conrad et al.

    Impact of the hypothalamic-pituitary-adrenal/gonadal axes on trajectory of age-related cognitive decline

    Prog. Brain Res

    (2010)
  • J.M. Daniel et al.

    Estrogen enhances performance of female rats during acquisition of a radial arm maze

    Horm. Behav.

    (1997)
  • J.M. Daniel et al.

    Effects of ovarian hormones and environment on radial maze and water maze performance of female rats

    Physiol. Behav

    (1999)
  • J.M. Daniel et al.

    Role of hippocampal M2 muscarinic receptors in the estrogen-induced enhancement of working memory

    Neuroscience

    (2005)
  • J.M. Daniel et al.

    Estrogen increases the sensitivity of ovariectomized rats to the disruptive effects produced by antagonism of D2 but not D1 dopamine receptors during performance of a response learning task

    Horm. Behav

    (2006)
  • J.L. Downs et al.

    The role of the brain in female reproductive aging

    Mol Cell Endocrinol.

    (2009)
  • E. Engler-Chiurazzi et al.

    Tonic Premarin dose-dependently enhances memory, affects neurotrophin protein levels and alters gene expression in middle-aged rats

    Neurobiol. Aging.

    (2011)
  • E.B. Engler-Chiurazzi et al.

    Continuous estrone treatment impairs spatial memory and does not impact number of basal forebrain cholinergic neurons in the surgically menopausal middle-aged rat

    Horm Behav.

    (2012)
  • T.C. Foster et al.

    Interaction of age and chronic estradiol replacement on memory and markers of brain aging

    Neurobiol. Aging.

    (2003)
  • T.C. Foster et al.

    Viral vector-mediated delivery of estrogen receptor-alpha to the hippocampus improves spatial learning in estrogen receptor-alpha knockout mice

    Mol. Ther

    (2008)
  • K.M. Frick et al.

    Estrogen replacement improves spatial reference memory and increases hippocampal synaptophysin in aged female mice

    Neuroscience

    (2002)
  • C.A. Frye et al.

    Estrogens and progestins enhance spatial learning of intact and ovariectomized rats in the object placement task

    Neurobiol. Learn. Mem

    (2007)
  • H.N. Fugger et al.

    Novel effects of estradiol and estrogen receptor alpha and beta on cognitive function

    Brain Res.

    (2000)
  • L.A. Galea et al.

    High levels of estradiol disrupt conditioned place preference learning, stimulus response learning and reference memory but have limited effects on working memory

    Behav. Brain Res.

    (2001)
  • R.B. Gibbs et al.

    Effects of estrogen replacement on the relative levels of choline acetyltransferase, trkA, and nerve growth factor messenger RNAs in the basal forebrain and hippocampal formation of adult rats

    Exp. Neurol

    (1994)
  • R.B. Gibbs

    Effects of gonadal hormone replacement on measures of basal forebrain cholinergic function

    Neuroscience

    (2000)
  • R.B. Gibbs

    Long-term treatment with estrogen and progesterone enhances acquisition of a spatial memory task by ovariectomized aged rats

    Neurobiol. Aging

    (2000)
  • R.B. Gibbs

    Basal forebrain cholinergic neurons are necessary for estrogen to enhance acquisition of a delayed matching-to-position T-maze task

    Horm. Behav

    (2002)
  • R.B. Gibbs

    Estradiol enhances DMP acquisition via a mechanism not mediated by turning strategy but which requires intact basal forebrain cholinergic projections

    Horm. Behav

    (2007)
  • R.B. Gibbs et al.

    Galanthamine plus estradiol treatment enhances cognitive performance in aged ovariectomized rats

    Horm. Behav.

    (2011)
  • R.B. Gibbs et al.

    Donepezil plus estradiol treatment enhances learning and delay-dependent memory performance by young ovariectomized rats with partial loss of septal cholinergic neurons

    Horm. Behav

    (2011)
  • E. Gomez-Gil et al.

    Androgen treatment effects on memory in female-to-male transsexuals

    Psychoneuroendocrinology

    (2009)
  • C. Gouchie et al.

    The relationship between testosterone levels and cognitive ability patterns

    Psychoneuroendocrinology

    (1991)
  • U. Halbreich et al.

    Possible acceleration of age effects on cognition following menopause

    J. Psychiatr. Res.

    (1995)
  • R. Hammond et al.

    Chronic treatment with estrogen receptor agonists restores acquisition of a spatial learning task in young ovariectomized rats

    Horm. Behav.

    (2009)
  • J.I. Acosta et al.

    Transitional versus surgical menopause in a rodent model: etiology of ovarian hormone loss impacts memory and the acetylcholine system

    Endocrinology

    (2009)
  • J.I. Acosta et al.

    The cognitive effects of conjugated equine estrogens depend on whether menopause etiology is transitional or surgical

    Endocrinology

    (2010)
  • M.M. Adams et al.

    Estrogen and aging affect the subcellular distribution of estrogen receptor-alpha in the hippocampus of female rats

    J. Neurosci.

    (2002)
  • J.P. Aggleton et al.

    Working memory in aged rats

    Behav. Neurosci.

    (1989)
  • C.C. Aguirre et al.

    Progesterone reverses 17beta-estradiol-mediated neuroprotection and BDNF induction in cultured hippocampal slices

    Eur. J. Neurosci

    (2009)
  • M. Alkondon et al.

    Choline and selective antagonists identify two subtypes of nicotinic acetylcholine receptors that modulate GABA release from CA1 interneurons in rat hippocampal slices

    J. Neurosci

    (1999)
  • Cited by (0)

    View full text